AU2009336567B2 - Anti-RhD monoclonal antibodies - Google Patents

Anti-RhD monoclonal antibodies Download PDF

Info

Publication number
AU2009336567B2
AU2009336567B2 AU2009336567A AU2009336567A AU2009336567B2 AU 2009336567 B2 AU2009336567 B2 AU 2009336567B2 AU 2009336567 A AU2009336567 A AU 2009336567A AU 2009336567 A AU2009336567 A AU 2009336567A AU 2009336567 B2 AU2009336567 B2 AU 2009336567B2
Authority
AU
Australia
Prior art keywords
cdrs
antibody
seq
variable region
rhd
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
AU2009336567A
Other versions
AU2009336567A1 (en
Inventor
Tomas Cinek
Gautam Vinod Daftary
John Kaundinya
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bharat Serums and Vaccines Ltd
Original Assignee
Bharat Serums and Vaccines Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bharat Serums and Vaccines Ltd filed Critical Bharat Serums and Vaccines Ltd
Publication of AU2009336567A1 publication Critical patent/AU2009336567A1/en
Priority to AU2014200308A priority Critical patent/AU2014200308B2/en
Application granted granted Critical
Publication of AU2009336567B2 publication Critical patent/AU2009336567B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/34Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against blood group antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]

Abstract

Anti-RhD monoclonal antibodies and methods for the production thereof.

Description

WO 2010/079510 PCT/IN2009/000741 ANTI-RHD MONOCLONAL ANTIBODIES Field of Invention The present invention relates to the production and use of anti-Rhesus D 5 mononclonal antibodies and antigen binding fragments thereof. Background and prior art Rhesus D antigen (also referred to in the art as RhD antigen, Rhesus factor, and/or Rh factor) is an antigen which may be present on the surface of 10 human red blood cells. Those individuals whose red blood cells have this antigen are usually referred to as "RhD-positive", while those individuals whose red blood cells do not have this antigen are referred to as "RhD-negative". A person who is RhD-negative and has never been exposed to the RhD 15 antigen will not produce .anti-RhD antibodies (antibdoies against the RhD antigen). However, transfer of RhD-positive blood to a RhD-negative individual will lead to sensitisation (immunization) of the RhD-negative individual against the RhD antigen. This can lead to a number of complications. In particular, where a RhD-negative woman gives birth to a RhD-positive infant there is a risk 20 of small amounts of the infant's blood entering the maternal circulation, causing the the mother to produce anti-RhD antibodies. Whilst this will not normally harm the first baby, should the now immunized mother fall pregnant with another RhD positive child then maternal anti-RhD antibodies may cross the placenta and attack the infant's blood cells, leading to a condition known as. haemolytic 25 disease of the newborn (HDN). Anti-RhD antibodies are therefore routinely administered to RhD negative patients where there is a risk of exposure to RhD-positive blood, in order to prevent the patient from becoming immunized against the RhD-positive 30 blood. For example, a RhD-negative patient may be given anti-RhD antibodies: prior to and/or shortly after giving birth to or having an abortion of an RhD-positve baby; after any incident during pregnancy which may have lead to bleeding across the placenta; as a routine preventative measure during pregnancy; or -1 - WO 20101079510 PCT/IN2009/000741 prior to or soon after any transfusion of blood components containing RhD positive red blood cells. Traditionally, the anti-RhD antibodies used have been polyclonal 5 antibodies obtained from the blood plasma of RhD negative volunteers who have been repeatedly immunized against RhD-positive red blood cells. However, the use of polyclonal antibodies has a number of recognised drawbacks, not least of which are the continuing need for a number of volunteer donors sufficient to meet the demand for antibody, and the risk of contamination of the antibody 10 preparation with any viruses or other pathogens that may be present in the donor's blood. Whereas polyclonal antibodies constitute antibodies secreted by a number of different plasma cells, and thus constitute a mixture of immunoglobulin 15- molecules secreted against a specific antigen and potentially recognising a variety of epitopes, monoclonal antibodies are produced from cells that are all clones of a single parent cell, and thus constitute a homogeneous population of antibodies, as is well known in the art. The cell lines from which monoclonal antibodies are produced are developed and cultured in-vitro, and this means 20 monoclonal antibodies have the potential to be produced as and when required both in large amounts and at high levels of purity. Accordingly, monoclonal anti RhD antibodies have a number of potential advantages over the polyclonal anti RhD antibody preparations that have traditionally been used. 25 A number of techniques for producing human monoclonal antibodies in general, and human monoclonal anti-RhD antibodies in particular, have been described. For example, EP-A2-0251440 discloses an anti-RhD monoclonal antibody producing heterohybridoma formed by fusion of non-Ig secreting mouse mylenoma cells with an anti-RhD Ig producing population of Epstein Barr virus 30 (EBV) transformed human lymphocytes. US 5,665,356 describes the production of human monoclonal anti-RhD antibodies having certain defined characteristics, produced by culturing selected EBV-transformed human B-lymphocytes. -2- WO 20101079510 PCT/IN2009/000741 US 6,312,690 describes the production anti-RhD monoclonal antibodies by recombinant techniques. An EBV immortalized human cell line producing an anti-Rhesus D monoclonal antibody called D7C2 was selected. The sequences encoding the variable regions of the heavy (H) and light (L) chains of D7C2 were 5 cloned, sequenced, and inserted into a recombinant baculovirus expression vector under the control of a strong baculovirus promoter. Insect cells transfected with the recombinant baculovirus were cultured, and the recombinant D7C2 monoclonal antibody recovered from the cell supernatant. 10 US-A1-2003/0175969 describes a method for preparing a anti-RhD monoclonal antibodies capable of activating effector cells expressing FcyRill, comprising: a) purifying monoclonal antibodies obtained from cell lines selected from human B lymphocyte heterohybridomas, or recombinant animal or human cell lines (such as CHO-K, CHO-Lec1 0, CHO Lec-1, CHO Pro-5, CHO dhfr-, Wil 15 2, Jurkat, Vero, Molt-4, COS-7, HEK293, YB2/0, BHK, K6H6, NSO, SP2/0-Ag 14 and P3X63Ag8.653 cells); b) adding each antibody obtained in step a) to a different reaction mixture comprising RhD-positive red blood cells, effector cells comprising cells expressing FcyRIll, polyvalent IgGs; and c) determining the percentage lysis of the target cells and selecting the monoclonal antibodies 20 which activate the effector cells causing significant lysis of the RhD-positive red blood cells. US 6,475,787 discloses a method for preparing monoclonal antibodies, in which a suitable eukaryotic host cell is transformed with a DNA sequence 25 encoding an antibody heavy chain and a DNA sequence encoding an antibody light chain, the two sequences being linked to different amplifiable marker genes so as to allow differential amplification of the heavy and light chain DNAs in order to optimize the relative gene copy numbers of the heavy and light chain DNAs. In a preferred embodiment the host cell is a Chinese Hamster Ovary (CHO) cell 30 which is DHFR deficient (i.e. incapable of producing dihydrofolate reductase), one of the amplifiable marker genes is an adenosine deaminase (ADA) gene, and the other is a DHFR gene. Amplification of the DNA encoding one antibody chain and linked in the ADA gene can then be achieved by treating the recombinant cells with increasing concentrations of 2'-deoxycoformycin, whilst -3amplification of the DNA encoding the other antibody chain and linked in the DHFR gene is achieved by treating the cell with increasing concentrations of methotrexate (MTX). 5 Nevertheless, there remains a need for further anti-RhD monoclonal antibodies and methods for the production thereof. Description of the Invention According to a first embodiment of the present invention there is provided 10 an isolated anti-RhD monoclonal antibody comprising: a) a heavy chain variable region having first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 2, and a light chain variable region having a first, second and third CDRs 15 which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 4; or b) a heavy chain variable region having first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 6, and 20 a light chain variable region having first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 8; wherein two CDRs are substantially identical if they have amino acid sequences that are at least 80% identical and/or differ in no more than 25 one amino acid. According to a second embodiment of the present invention there is provided an isolated polynucleotide encoding the light and/or heavy chain of an antibody according to the first embodiment. According to a third embodiment of the present invention there is 30 provided an expression vector including coding sequences encoding the light and heavy chains of an antibody according to the first embodiment. -4- According to a fourth embodiment of the present invention there is provided expression system including coding sequences encoding the light and heavy chains of an antibody according to the first embodiment, the expression 5 system comprising: a first expression vector including the coding sequence encoding the light chain; and a second expression vector including the coding sequence encoding the heavy chain. 10 According to a fifth embodiment of the present invention there is provided a cell transformed with the expression vector according to the third embodiment or the expression system according to the fourth embodiment. According to a sixth embodiment of the present invention there is provided a method of manufacturing monoclonal antibodies, comprising 15 cultivating recombinant cells according to the fifth embodiment, and recovering the monoclonal antibody from the culture medium. According to a seventh embodiment of the present invention there is provided a pharmaceutical composition comprising a monoclonal antibody according to the first embodiment, the isolated polynucleotide of the second 20 embodiment, the expression vector of the third embodiment, the expression system of the fifth embodiment or the cell of the sixth embodiment. According to an eighth embodiment of the present invention there is provided the use of a monoclonal antibody according to the first embodiment, the isolated polynucleotide of the second embodiment, the expression vector of the 25 third embodiment, the expression system of the fifth embodiment or the cell of the sixth embodiment in the manufacture of a medicament for inhibiting or preventing immunization of a RhD-negative human patient against RhD-positive blood. According to a first aspect of the present invention there is provided an 30 isolated anti-RhD monoclonal antibody comprising: a) a heavy chain variable region having first, second and third CDRs (complementarity determining regions) which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 2, and a - 4a light chain variable region having first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 4; or b) a heavy chain variable region having first, second and third CDRs 5 which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 6, and a light chain variable region having first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 8; or c) a heavy chain variable region having first, second and third CDRs 10 which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 10, and a light chain variable region having first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 12. It is to be noted that, throughout the description and claims of this 15 specification, the word 'comprise' and variations of the word, such as 'comprising' and 'comprises', is not intended to exclude other variants or additional components, integers or steps. Modifications and improvements to the invention will be readily apparent to those skilled in the art. Such modifications and improvements are intended to be within the scope of this invention. 20 Any reference to or discussion of any document, act or item of knowledge in this specification is included solely for the purpose of providing a context for the present invention. It is not suggested or represented that any of these matters or any combination thereof formed at the priority date part of the common general knowledge, or was known to be relevant to an attempt to solve 25 any problem with which this specification is concerned. As used herein, the term "anti-RhD antibody" refers to both whole antibodies and to fragments thereof that have binding specificity for RhD antigen. The binding affinity/specificity of an antibody can be measured by a various assays, as will be known to and can be routinely implemented by one of ordinary 30 skill in the art. For example, antibodies recognising and specifically binding to RhD antigen can be determined using one or more standard techniques as - 4b - WO 20101079510 PCT/IN2009/000741 known to one of ordinary skill in the art, such as but not limited to: EIA / ELISA techniques, such as competitive EIA (enzyme linked-immunoassay); flow cytometry; and/or ADCC (antibody-dependant cellular toxicity) assays. Exemplary competitive EIA, flow cytometry, and ADCC techniques are described 5 in further detail in the Examples that follow. As is well known in the art, whole antibodies are typically formed of one or two heavy and one or two light chains. The heavy and light chains each comprise a variable region and a constant region. The variable regions (also 10 referred to as the variable domains) dictate the antibody's antigen binding specificity. Each variable domain is composed of complementarity determining regions (CDRs, of which there are typically three, designated CDR1, CDR2 and CDR3) interspersed with more conserved regions known as framework regions. On folding of the antibody to adopt the correct quaternary structure, the CDRs of 15 a heavy and light chain together form the antigen binding site. The constant region of the heavy chain is composed of three or more constant domains and is dependent on the class (eg. IgA, IgD, IgE, IgG, or igM) and isotype (eg. IgAl, IgA2, IgG1, IgG2, IgG3, IgG4) of the antibody. It is identical in all antibodies of the same class and isotype, but differs in antibodies of different isotypes. The 20 light chain constant region is composed of a single constant domain of which is of one of two isotypes, kappa or lambda, and is likewise identical in all antibodies of the same isotype. The constant regions of the antibodies typically mediate binding of the antibody to host tissues or factors. 25 Antibody fragments according to the present invention typically include at least the CDRs and sufficient of the framework regions to specifically bind the antigen. Exemplary types of fragment include, but are not limited to, a Fab' fragment (consisting of the variable domain and a constant domain of both the light and heavy chains), a F(ab')2 fragment (two Fab' fragments linked by a 30 disulfide bridge at the hinge region), a Fv fragment (consisting of the variable domains only of the light and heavy chains), and other types of fragment as known to one skilled in the art. -5- WO 20101079510 PCT/IN2009/000741 SEQ ID NOs: 2 and 4 are the amino acid sequences of the heavy and light chains of the anti-RhD monoclonal antibody referred to herein as RhD1 and described below in further detail. SEQ ID NOs: 6 and 8 are the amino acid sequences of the heavy and light chains of the anti-RhD monoclonal antibody 5 referred to herein as RhD2 and described below in further detail. SEQ ID NOs: 10 and 12 are the amino acid sequences of the heavy and light chains of the anti-RhD monoclonal antibody referred to herein as RhD3 and described below in further detail. 10 The antibodies according to the first aspect of the present invention therefore comprise heavy chain and light chain variable regions having first second and third complementarity determining regions (i.e. CDR1, CDR2 and CDR3) which are identical or substantially identical to the first second and third complementarity determining regions (CDR1, CDR2 and CDR3) of antibody 15 RhD1, RhD2 or RhD3. As used herein, two CDRs are "substantially identical" if they have amino acid sequences that preferably are at least 80% identical and/or differ in no more than one amino acid. More preferably the sequences are at least 90% identical 20 and/or differ in no more than one amino acid. Preferably, where amino acid substitutions occur such substitutions are conservative substitutions. Where the CDRs of two antibodies are at least substantially identical, it is reasonable to predict that the resulting antigen binding site of the two antibodies will have similar antigen binding properties. For example, antibodies RhD1 and RhD2 25 have highly similar CDRs, as can be seen from Figures 1 and 2 (described below in further detail), and both have high binding affinity for the RhD antigen. Most preferably, the CDRs of the antibody are identical to those of RhD1, RhD2 or RhD3. 30 As used herein the term "an isolated monoclonal antibody" refers to an antibody which has been produced by monoclonal techniques and which has been isolated from antibodies of other types. In other words, the only other antibodies present will be antibodies produced by cells of the same cell line (i.e. -6- WO 20101079510 PCT/IN2009/000741 cells all originating from the same single parent cell) as the cell which produced the monoclonal antibody. This is of course in contrast to, for example, polyclonal antibodies where the antibodies constitute a mixture of different antibodies originating from different plasma cells. 5 In a preferred embodiment, the isolated anti-RhD monoclonal antibody comprises heavy and light chain variable regions which are at least 80%, more preferably at least 90%, more preferably at least 95%, more preferably at least 98%, most preferably 100% identical to the respective variable regions of the 10 heavy and light chains of the RhD1, RhD2 or RhD3 antibody to which its CDRs are at least substantially identical. Thus, in this embodiment the antibody comprises either: a) a heavy chain variable region which is at least 80%, 90%, 95%, 15 98%, or 100% identical to the variable region of SEQ ID NO: 2 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 2, and a light chain variable region which is at least 80%, 90%, 95%, 98%, or 100% identical to the variable region of SEQ ID NO: 4 and has first, second and third CDRs which are 20 identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 4; or b) a heavy chain variable region which is at least 80%, 90%, 95%, 98%, or 100% identical to the variable region of SEQ ID NO: 6 and has first, 25 second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 6, and a light chain variable region which is at least 80%, 90%, 95%, 98%, or 100% identical to the variable region of SEQ ID NO: 8 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs 30 of SEQ ID NO: 8; or c) a heavy chain variable region which is at least 80%, 90%, 95%, 98%, or 100% identical to the variable region of SEQ ID NO: 10 and has first, second and third CDRs which are identical or substantially identical to the -7- WO 20101079510 PCT/IN2009/000741 respective first, second, and third CDRs of SEQ ID NO: 10, and a light chain variable region which is at least 80%, 90%, 95%, 98%, or 100% identical to the variable region of SEQ ID NO: 12 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third 5 CDRs of SEQ ID NO: 12. Techniques for identifying antibody variable regions and CDRs, comparing and aligning amino acid sequences, and determining the % identity between two amino acid sequences are well known in the art. For example, the 10 CDRs, variable regions, and constant regions of an antibody can be determined using software such as IMGTN-QUEST tool (http://imqt.cines.fr/IMGT vquest/share/textes/) using default settings, and/or via comparison with databases of known immunoglobulin sequences such as IMGT/GENE-DB (http://imqt.cines.fr/iMGT GENE-DB/GENElect?livret=0) or 15 V-BASE (http://vbase.mrc-cpe.cam.ac.ukl). Amino acid or nucleic acid sequence sequences, whether for whole antibodies or specific parts thereof, can be aligned and their % identity determined using ClustalW (http://www.ebi.ac.uk/Tools/clustalw/), ClustalW2 (http://www.ebi.ac.uk/fools/clustalw2) or GAP 20 (http://qenome.cs.mtu.edu/align/align.html) using default parameters, or using proprietary software such as Vector NTI v.10.3. In a preferred embodiment, the antibody further comprises a light chain constant domain and at least one heavy chain constant domain. The light chain 25 constant domain may be of either the kappa or lambda type. The heavy chain constant domain is preferably an IgG class constant domain. Thus, in this embodiment the antibody may for example be a Fab' or F(ab')2 fragment, as discussed above, or it may be a whole antibody. If the latter, preferably all the heavy chain constant domains are IgG domains (i.e. the antibody comprises an 30 IgG heavy chain constant region). In a particularly preferred embodiment the constant domain or region is an IgG 1 or IgG 3 constant domain or region. Preferably all constant domains (both light and heavy) are human constant domains. -8- WO 20101079510 PCT/IN2009/000741 According to a second aspect of the present invention, there is provided an isolated polynucleotide encoding the light and/or heavy chain of an antibody according to the first aspect. 5 As used herein, the term an "isolated polynucleotide" refers to a polynucleotide that has been isolated from a cellular environment (i.e. it is not present in a cell or organism), and it can be in purified form (i.e. substantially free of other polynucleotides, proteins, and cellular components) of form part of composition containing other polynucleotides and/or compounds. The term 10 "encoding a light chain" refers not only to sequences encoding whole light chains, but also to sequences encoding fragments thereof (such as the variable domain only) where the antibody to be expressed is an antibody fragment as described above. Similarly, the term "encoding a heavy chain" refers not only to sequences encoding whole heavy chains, but also to sequences encoding 15 fragments thereof (such as the variable domain only or the variable domain plus one or more but not all of constant domains) where the antibody to be expressed is an antibody fragment as described above. Exemplary nucleic acid sequences include the relevant coding sequences of 20 SEQ ID NOs: 1, 3, 5, 7, 9, and 11, which sequences are the coding sequences for, respectively, amino acid SEQ ID NOs: 2, 4, 6, 8, 10, and 12. Thus, for example, if the antibody comprises identical variable regions to the variable regions of SEQ ID NOs: 2 and 4 (the heavy and light chains of the anti-RhD antibody designated RhD 1), then an exemplary nucleic acid sequence could 25 comprise the sections of SEQ ID NOs: 1 and 3 that encode said variable regions. Alternatively, such nucleic acid sequences could be modified for optimised expression (i.e. transcription and/or translation) in the desired host cell, for example via techniques known to one of skill in the art. For example, optimization of the native nucleic acid sequence may comprise one or more of: 30 optimizing the GC distribution, and AT/GC stretches (to enhance the stability of mRNA); removing inhibitory motifs (such as premature polyA signals); removing cryptic splice sites (to prevent alternative, incorrect splicing of mRNA); optimizing mRNA secondary structure (to avoid tight hairpins possibly stalling translation); optimizing open reading frames (to avoid secondary or alternative reading -9- WO 20101079510 PCT/IN2009/000741 frames); and optimizing codon usage (to avoid rare codons that can slow down translation). According to a third aspect of the present invention, there is provided an 5 expression system comprising one or more expression vectors and including coding sequences encoding the light and heavy chains of an antibody according to the first aspect. The expression vector(s) may be of any type used in the art, such as for 10 example plasmids and viral vectors. The expression vectors of the present invention are preferably plasmids. In addition to the antibody chain coding sequences, the vector(s) will include the necessary regulatory sequences for proper transcription and translation of the coding sequences in the intended host cell, such as for example a suitable promoter and polyadenylation (polyA) 15 sequence. The vector(s) may further comprise a Kozak sequence for increased efficiency of expression, and/or a sequence encoding for a signal peptide for post translational transport of the antibody chains (for example for secretion of the antibodies). A further preferred feature is the presence of one or more antibiotic resistance genes and/or other forms of selection marker, allowing for selection of 20 cells that have been stably transfected with. the vector, and/or that display stronger expression of the antibody coding sequences, as discussed below in more detail. The promters and poly(A) sequences used to drive expression of the light 25 and heavy chain coding sequences may be of any type used in the art. A variety of different promoters and poly(A) seqences are known, the selection of appropriate promoters and poly(A) sequences for use in the chosen host cell being well within the abilities of one of ordinary skill in the art. For example, suitable promoters for use in a mammalian host cell include the SV40 early and 30 late, elgongation factor 1 (EF-1), and cytomegalovirus (CMV) promoters. Suitable poly(A) sequences include those from SV40 poly(A), bovine growth hormone (BGH), thymidin kinase (TK), and human growth hormone (hGH). In a preferred embodiment, the light and heavy chain coding sequences are driven by - 10- WO 20101079510 PCT/IN2009/000741 the human elongation factor 1 alpha (hEF-la) promoter and BGH poly(A) sequence. In one embodiment, the expression system comprises an expression 5 vector that includes both the coding sequence for the light chain and the coding sequence for the heavy chain. In an alternative embodiment, the light and heavy chain coding sequences are carried by separate vectors, the expression system comprising: 10 a first expression vector including the coding sequence encoding the light chain; and a second expression vector including the coding sequence encoding the heavy chain. 15 In this embodiment, one or both of said first and second expression vectors may include a dihydrofolate reductase (dhfr) selection marker. This marker comprises a coding sequence for DHFR, which is coupled to suitable promoter and polyadenylation sequences, preferably the SV40 early (SV40E) promoter and poly(A) sequences. DHFR allows de novo synthesis of the DNA 20 precursor thymidine. Therefore, by transfecting a host cell-line which is DHFR deficient (i.e. which is itself incapable of producing DHFR), one can then select for cells which have stably integrated the vector into their genome by growing the cells in a medium deficient in deoxyribonucleosides and ribonucleosides. Moreover, once the successfully transfected cells have been isolated, the 25 expression of the desired coding sequence(s) (i.e. the light and/or heavy chain) can be amplified by using the DHFR inhibitor methotrexate (MTX), which causes some cells to react by amplifying large regions of DNA surrounding the dhfr gene. 30 In a preferred embodiment, one of said first and second expression vectors includes an antibiotic resistance gene (a nucleic acid sequence that imparts resistance to the antibiotic in question) but does not include the DHFR coding sequence, and the other of said expression vectors includes the DHFR coding sequence but does not include a gene providing resistance to the same -11 - WO 20101079510 PCT/IN2009/000741 antibiotic as said antibiotic resistance gene. The antibiotic resistance gene may be of any type used in the art. For example, suitable antibiotic resistance genes for imparting resistance to a mammalian host cell include: aminoglycoside (e.g. neomycin, hygromycin B) resistance genes, such as neomycin 5 phosphotransferase (npt) and hygromycin B phosphotransferase (hpt, hph); aminonucleoside (eg. puromycin) resistance genes such as puromycin N acetyltransferase (pac); glycopeptide (eg. bleomycin, phleomycin) resistance genes such as the ble gene; and peptidyl nucleoside (eg. blasticidin) resistance genes such as the b/s, bsr or bsd genes. As with the dhfr selection marker, the 10 antibiotic resistance gene may as needed be coupled to any suitable promoter and polyadenylation sequences. Preferred are the SV40 early (SV40E) promoter and poly(A) sequences. In a particularly preferred embodiment, the antibiotic resistance gene 15 comprises a neomycin phosphotransferase (NPT) coding sequence. The cells stably transfected with the vector including the NPT coding sequence can then be selected for by growing the cells in a medium containing neomycin, or a neomycin analog such as G418, the toxic effects of which are neutralized by NPT. 20 Thus, the above described embodiment, in which one vector has the dhfr selection marker and the other has the antibiotic selection gene, allows for selection of only those cells which have stably integrated both vectors into their genome by growing the cells in a medium deficient in deoxyribonucleosides and 25 ribonucleosides and containing the relevant antibiotic (such as neomycin or a suitable analogue where the antibiotic resistance gene is the npt gene). Cells that were not transfected or were transfected with only one plasmid will not survive the selection process. Moreover, because the co-transfected plasmids often integrate into one spot of the genome, subsequent growth of the 30 successfully transfected cells in increasing concentrations of MTX can still be used to effectively amplify expression of the antibody chains encoded by both vectors (i.e. to amplify expression of both the heavy and light chain sequences). - 12- WO 20101079510 PCT/IN2009/000741 It should be noted that while, in this embodiment, the vector carrying the dhfr selection marker does not include a gene providing resistance to the same antibiotic as the antibiotic resistance gene carried by the other vector, it and indeed both vectors may further comprise a different antibiotic resistance gene 5 providing resistance against a further antibiotic. Again, the additional antibiotic gene may be of any type used in the art. For example, where one but not both vectors carries an NPT coding sequence (providing resistance against neomycin and analogues thereof) both vectors may usefully additionally comprise an ampicillin resistance (AmpR) gene, for the purpose of providing ampicillin 10 resistance when incorporated into a bacterial host cell. Other antibiotic resistance genes that are commonly used to impart resistance in bacterial hosts include: plactamase genes (providing resistance to plactam antibiotics such as ampicillin and other penicillins), such as TEM-1 &-lactamase; genes providing resistance to aminoglycosides such as streptomycin, kanamycin, tobramycin, 15 and amikacin; and tetracycline (e.g. tetracycline, doxycycline, minocycline, oxtetracycline) resistance genes, such as the tetA genes. According to a fourth aspect, the present invention provides a cell transformed with an expression system according to the third aspect or fourth 20 aspects. The host cells for use in the present invention may be of any suitable type. However, in a preferred embodiment the host cell (cell to be transfected) is a eukaryotic cell, more preferably a vertebrate cell, most preferably a mammalian 25 cell. A variety of suitable mammalian host cells are available, such as are for example listed in US-A1-2003/0175969 referred to above. Preferred mammalian host cells include: all variants of CHO cells, such as CHO K1 and dhfr-deficient CHO (DG44, DXB11); HEK293; BHK; COS-1 and COS-7; NSO; and PER.C6. The preferred host cells are Chinese Hamster Ovary (CHO) cells, in particular 30 dhfr-deficient CHO cells (dfhr- CHO cells). The host cells may be transfected with the expression vectors using standard techniques and transfection conditions, such as are known in the art. Exemplary transfection conditions are provided in the Examples that follow. -13- WO 20101079510 PCT/IN2009/000741 According to a fifth aspect, the present invention provides a method of manufacturing monoclonal antibodies, comprising cultivating recombinant cells according to the fourth aspect, and recovering the monoclonal antibody from the culture medium. Exemplary growth media and conditions are provided in the 5 Examples that follow, but any suitable growth conditions and commercial or custom growth media can be used, as are routinely employed in the art. Likewise, any standard technique for purifying secreted antibodies from growth media can be employed, exemplary techniques being again outlined below. 10 According to a sixth aspect, the present invention provides a pharmaceutical composition comprising a monoclonal antibody according to the first aspect. Preferably, the pharmaceutical composition also comprises a pharmaceutically acceptable carrier. 15 The monoclonal antibodies can be formulated as desired dependent on the intended route of administration. For example, the monoclonal antibodies may be formulated for injection (for example intra-muscularly) analogous to conventional polyclonal anti-D formulations. Exemplary dosages range from 150 to 300 micrograms (as measured by agglutination titer, as described below in 20 further detail). Exemplary carriers include: phosphate-buffered saline; and glycine saline buffer. The composition may comprise monoclonal antibodies of a single type only (i.e. the only antibodies present in the composition are antibodies produced 25 by cells of the same cell line). Alternatively, the composition may comprise a combination of more than one type of monoclonal antibody. For example, the composition could comprise two or more distinct types of monoclonal antibodies that are in accordance with the first aspect of the invention, such as a combination of two or all three of monoclonal antibodies RhD1, RhD2 and/or 30 RhD3. Alternatively or additionally, the composition could comprise, in addition to monoclonal antibodies according to the first aspect of the present invention, other anti-RhD monoclonal antibodies as for example are known from the art. In a preferred embodiment, the composition comprises at least one monoclonal - 14- WO 20101079510 PCT/IN2009/000741 antibody that has an lgG 1 constant domain or region, and at least one monoclonal antibody that has an igG 3 constant domain or region. Where the composition comprises a combination of more than one type 5 of 'monoclonal antibody, it is preferred that the composition comprises no more than 50 different types of monoclonal antibody. More preferably, the composition comprises at most 25, 20, 15, 10 or 5 different types. According to a seventh aspect, the present invention provides a method 10 of inhibiting or preventing immunization of a RhD-negative human patient against RhD-positive blood, comprising administering a prophylactically effective amount of a monoclonal antibody according to the first aspect or pharmaceutical composition according to the sixth aspect. 15 Specific indications and/or circumstances in which the monoclonal antibodies may be administered correspond to those for which the existing anti RhD polyclonal antibodies are administered. According to an eighth aspect, the present invention provides a 20 monoclonal antibody according to the first aspect, or a pharmaceutical composition according to the sixth aspect, for use in a method of inhibiting or preventing immunization of a RhD-negative human patient against RhD-positive blood. 25 According to a ninth aspect, the present invention provides the use of a monoclonal antibody according to the first aspect in the manufacture of a medicament for inhibiting or preventing immunization of a RhD-negative human patient against RhD-positive blood. 30 The invention is further illustrated in the following non-limiting Examples, with reference also to the accompanying drawings in which: -15- WO 20101079510 PCT/IN2009/000741 Figure 1 is an alignment of amino acid sequences of the heavy chains of monoclonal antibodies RhD1, RhD2 and RhD3, in which the variable regions have been underlined and the complementarity determining regions highlighted in bold and shaded; 5 Figure 2 is an alignment of amino acid sequences of the light chains of monoclonal antibodies RhD1, RhD2 and RhD3, in which the variable regions have been underlined and the complementarity determining regions highlighted in bold; 10 Figure 3 is a map of plasmid vector pCB3; Figure 4 is a map of plasmid vector pCB11; 15 Figure 5 is a map of pCB3 containing an anti-RhD antibody heavy chain (RhD HC) coding sequence; and Figure 6 is a map of pCB11 containing an anti-RhD antibody light chain (RhD LC) coding sequence; 20 Figure 7 is an example of a dose-response curve generated in an ADCC assay, in which cytotoxicity is plotted against the logarithm of antibody concentration at which the erythrocytes were presensitized; and 25 Figure 8 is an example of linear regression performed on the relevant data points taken from Fig. 7. Sequence listings which are 48 in number are provided after the Drawings. 30 The Sequence listings are also provided separately in accompanying CD in electronic form. - 16 - WO 20101079510 PCT/IN2009/000741 Examples Isolation of peripheral blood mononuclear cells (PBMCs) and B cells from peripheral blood of healthy volunteers hyperimmunized with Rhesus D 5 (RhD)-positive red blood cells Blood from healthy RhD-negative volunteers repeatedly immunized with red blood cells isolated from healthy RhD-positive individuals of the same ABO blood group was sourced from Cliniqa. Within four weeks after the last 10 immunization the anti-RhD titer in serum was checked, the volunteers were bled, their peripheral blood mononuclear cells (PBMCs) were separated from other blood cell populations by Ficoll-Hypaque (Pharmacia) gradient centrifugation, and the cells were either used fresh or cryopreserved for later use. T cells were routinely depleted by rosetting with 2% S-(2-Aminoethyl)isothiouronium bromide 15 hydrobromide (AET)-treated sheep red blood cells and the resulting enriched B cells were transformed by Epstein-Barr virus (EBV). EBV transformation 20 Since EBV activation has been shown to be advantageous for subsequent fusion of human B cells with the respective fusion partner, enriched B cells were transformed by EBV using spent supernatant from the B95-8 marmoset cell line as a source of the virus. The B cells resuspended in a complete IMDM medium (Gibco) with 30% fetal calf serum (FCS) were seeded in 25 96-well plates at a concentration between 5x1 03 and 2.5x1 04 cells/well. The B95-8 supernatant was added to the wells in an amount ranging from 5% to 40% of the total volume. The plates were incubated in a humidified 5% CO 2 incubator at 37 0 C for two to four weeks before screening. 30 Screening of plates for transformants secreting anti-RhD antibodies Supernatants of transformed B cells were screened for the presence of anti-RhD antibodies by competitive enzyme-linked immunoassay (EIA). The principle of the test is as follows: a labeled monoclonal anti-RhD reference -17- WO 20101079510 PCT/IN2009/000741 antibody of known binding affinity and specificity (Brad-5; NIBSC) competes with an unlabeled antibody (in this case, the secreted antibodies in the supernatants) for binding to RhD-positive erythrocytes. An inhibition of the reference monoclonal antibody (mAb) binding indicates the presence of RhD-specific 5 antibodies that bind to the same immunodominant epitope as the reference mAb. The degree of inhibition of the reference mAb binding correlates to the concentration and affinity of the interfering antibodies. RhD-positive erythrocytes (R2R2 haplotype; ImmucorGamma) treated 10 with papain were fixed with glutaraldehyde and immobilized on the bottom of 96 well flat-bottom test plates. After extensive washing and blocking of the plates, the supernatants from transformed B cells, the standards, and negative controls were added to the wells and the plates were incubated for 30-60 min at room temperature (RT). The plates were washed three times. The biotinylated 15 reference mAb was added and the plates were incubated for 30 more minutes at RT. The plates were washed again and incubated with a secondary reagent, ExtrAvidin-Alkaline Phosphatase conjugate (Sigma) for 30 min at RT. After another washing step, Sigma Fast PNPP (p-Nitrophenyl Phosphate) substrate (Sigma) was added. When the color developed sufficiently, the reaction was 20 stopped with 3N NaOH and the binding of the reference mAb was detected by reading the optical densities (at 405nm) on a plate reader (Bio-Rad). The data was analyzed with a software package supplied with the plate reader. Cell fusion 25 Because human B cells transformed with EBV are unstable and can rapidly cease to produce antibodies, fusion with a suitable fusion partner is usually necessary to prolong their lifetime and enable their subcloning. Therefore, any cultures of transformed B cells that produced antibodies inhibiting 30 binding of the biotinylated reference antibody to RhD* erythrocytes as assessed by EIA (see above) were fused to a human heterohybridoma K 6
H
6 /B5 either by the standard polyethylene glycol (PEG) method or by electrofusion. The electrofusion was performed with the electrofusion apparatus (Eppendorf - 18- WO 20101079510 PCT/IN2009/000741 Multiporator) and an electrofusion buffer (Eppendorf) according to manufacturer's protocols. Subcloning of hybridomas 5 Subclones were grown on feeder layers established from newborn foreskin fibroblast line CCD-1114Sk (ATCC). Feeders were maintained in IMDM media containing 2-20% fetal bovine serum (FBS), depending on cell growth. Feeder trays were treated with UV light on the day of subcloning. The cell lines 10 to be subcloned were counted, the appropriate dilutions to plate approximately 0.3 cells/well were prepared, and the cell suspensions were. pipetted into the 96 well plates containing the feeder layer. Each cell line was seeded in at least two plates. The cultures were fed every 3-4 days. The supernatants from wells exhibiting growth of hybridomas were tested by EIA usually in 3-4 weeks. 15 Hybridoma clones selected for development of recombinant cell lines Hybridoma clones selected for development of recombinant antibodies are listed in Table 1 (below). Each clone was assigned a simplified designation 20 for the purpose of recombinant cell line development. Table 1. Designation of Anti-RhD Antibodies Hybridoma clone: Antibody isotype: Clone designation: 25 SD30.06.F5.1G2 human IgG1, lambda RhD1 SD30.02.C3.3D11 human IgG1, lambda RhD2 SD412.04.G11.2D10 human IgG3, kappa RhD3 30 RNA isolation Total RNA from the hybridoma cells was purified using Trizol reagent (Invitrogen) according to the protocol suggested by the manufacturer with the additional step of RNA extraction with chloroform to remove traces of phenol. - 19- WO 20101079510 PCT/IN2009/000741 Spectrophotometrical RNA quantification was carried out at 260 nm assuming 1 OD to be equivalent to 40 ug/ml RNA. First strand synthesis 5 The first strand of cDNA was synthesized using the Super Script Ill First Strand System for RT-PCR (Invitrogen) according to the protocol suggested by the supplier. Oligo d(T) primer from the kit was used in all cases to prime the reactions. 10 RNA hydrolysis The removal of RNA molecules from reverse transcription reaction was 15 carried out by RNaseH digestion (Super Script Ill First-Strand System for RT PCR) according to manufacturer's instructions. First-strand cDNA was cleaned using QlAquick PCR Purification Kit (Qiagen). Tailing of first-strand cDNA 20 To facilitate amplification of first-strand cDNA with unknown 3' sequence, poly(A) tail was appended to the 3' end of each cDNA to create a defined. priming site. For this purpose, recombinant Terminal Deoxynucleotidyl Transferase (Invitrogen) was used. The reaction was carried out according to manufacturer's 25 recommendations. Reaction product was cleaned using QlAquick PCR Purification Kit (Qiagen). PCR amplification of Ig heavy- (HCs) and light chains (LCs) 30 The primers (SEQ ID NOs: 13 to 19) used for PCR amplification of the heavy and light chain coding sequences from the first-strand cDNA are listed below (EcoRI restriction sequence in each primer is underlined). - 20 - WO 20101079510 PCT/IN2009/000741 Forward primer (compatible with the poly(A) extension of the first strand of cDNA): For all chains: 5 5'-GACTGAATTC I I I I I T i I I I I II I I I V-3' Reverse primers (gene specific): For gamma chains: 10 5'-ACTGGAATTCGGTGCTTTATTTCCATGCTGG-3' 5'-ACTGGAATTCGTACGTGCCAAGCATCCTCG-3' For kappa chains: 5'-ACTGGAATTCAGAGGCCAAAGGATGGGAGG-3' 15 5'-GACTGAATTCCTGGAACTGAGGAGCAGGTGG-3' For lambda chains: 5'-GACTGAATTCCCTGGGATCCTGCAGCTC-3' 5'-ACTGGAATTCGGGGTGAGGGTTGAGAACC-3' 20 PCR was carried out using PfuUltra High-Fidelity thermostable DNA polymerase (Stratagene). Typically the first five cycles were primed only with the forward primer; annealing temperature was 45 0 C. After that, the reverse, gene specific primer was added and the PCR was extended for another 30-35 cycles 25 at annealing temperature of 50-65*C. Resulting fragments were gel purified using QlAquick Gel Extraction Kit (Qiagen), subcloned into pBluescript cloning vector and sequenced. Subcloning of PCR Products into pBluescript cloning vector 30 The purified PCR products were ligated using the Quick Ligation Kit (NEB) into pBluescript cloning vector (Stratagene) cut with EcoRV. DH5a bacterial cells were transformed with the resulting DNA and spread onto LB plates supplemented with 40pg/ml ampicillin and pre-treated with 50I of -21- WO 20101079510 PCT/IN2009/000741 20mg/mi Xgal and 25pl of 200mg/ml Isopropyl P-D-1-thiogalactopyranoside (IPTG). Colonies were blue/white selected for the presence of an insert. Isolation of Plasmid DNA and Sequencing 5 Selected white colonies were picked and expanded. The DNA was isolated with QiAprep Spin Miniprep Kit (Qiagen). A control digest was performed with EcoRI (both forward and reverse PCR primers contained an EcoRI site). Inserts in plasmids yielding the expected digestion pattern were 10 sequenced (Biotech Core). RhD1, RhD2 and RhD3 coding and amino acid sequences The amino acid sequences of the heavy chain (HC) and light chain (LC) 15 of RhD1, RhD2 and RhD3, and the corresponding nucleotide sequences encoding said heavy and light chains are set out in the accompanying sequence listing, as further explained below. The sequences were analyzed with the help of IMGT databases and 20 software (imgt.cines.fr). More specifically: the sequences of constant regions were determined from the IMGT/GENE-DB database of genomic Ig sequences (http://imqt.cines.fr/I MGT GEN E-DB/GENElect?livret=0), by selecting the species, locus, gene type, group (skipped subgroup) and functionality (e.g. 25 species: Homo sapiens, locus: IGH, gene type: constant, group: IGHC, functionality: functional), and searching the database - from the resulting list, the desired isotype (e.g. IGG1) was selected in order to identify appropriate IMGT/LIGM-DB reference sequence(s) for comparison with the RhD sequence; the variable regions were determined by subtracting the constant regions; 30 and the CDRs were determined using IMGTN-QUEST tool (http://imqt.cines.fr/IMGT vquest/share/textes/), by selecting the immunoglobulin species (human), uploading the nucleotide sequence of the complete antibody -22- WO 2010/079510 PCT/IN2009/000741 chain, or just its variable region, in FASTA format, and analyzing the sequence using IMGTN-QUEST default settings. For further information on IMGTN-QUEST tool and IMGT/GENE-DB see 5 also: Lefranc M.-P., Giudicelli V., Kaas Q., Duprat E., Jabado-Michaloud J., Scaviner D., Ginestoux C., Clement 0., Chaume D. and Lefranc G. IMGT, the international ImMunoGeneTics information system. Nucl. Acids Res., 2005, 33, D593-D597; 10 Giudicelli V., Chaume D. and Lefranc M.-P. IMGTN-QUEST, an integrated software for immunoglobulin and T cell receptor V-J and V-D-J rearrangement analysis. Nucl. Acids Res. 2004, 32, W435-W440; and, Giudicelli V., Chaume D. and Lefranc M.-P. IMGT/GENE-DB: a comprehensive database for human and mouse immunoglobulin and T cell 15 receptor genes. Nucl. Acids Res. 2005, 33, D256-D261. V-BASE (a database of all human germline variable region sequences; http://vbase.mrc-cpe.cam.ac.uk/) can also be used to determine, or corroborate, the ends of a variable region. Under Alignments, one can find germline 20 sequences of the signal peptides, V-segments, D-segments (if applicable), and J-segments of all heavy and light chains. It will be apparent from the IMGT analysis what segments are employed in a given antibody chain. One can then reference the particular J-segment in V-BASE to determine the exact ending. 25 SEQ ID NO: 1 is the nucleotide sequence of the coding region of RhD1 HC. Nucleotides 1-57 encode the signal peptide. Nucleotides 58-448 encode the variable region, of which nucleotides 133-156 encode CDR1, nucleotides 208-231 encode CDR2, and nucleotides 346-414 encode CDR3. Nucleotides 449-1437 encode the constant region (this being a gammal, or IgG1, constant 30 region). The amino acid sequence of RhD1 HC is given as SEQ ID NO: 2. SEQ ID NO: 3 is the nucleotide sequence of the coding region of RhD1 LC. Nucleotides 1-57 encode the signal peptide. Nucleotides 58-388 encode the variable region, of which nucleotides 133-159 encode CDR1, nucleotides -23- WO 20101079510 PCT/IN2009/000741 211-219 encode CDR2, and nucleotides 328-357 encode CDR3. Nucleotides 389-705 encode the constant region (this being a lambda constant region). The amino acid sequence of RhD1 LC is given as SEQ ID NO: 4. 5 SEQ ID NO: 5 is the nucleotide sequence of the coding region of RhD2 HC. Nucleotides 1-57 encode the signal peptide. Nucleotides 58-448 encode the variable region, of which nucleotides 133-156 encode CDR1, nucleotides 208-231 encode CDR2, and nucleotides 346-414 encode CDR3. Nucleotides 449-1437 encode the constant region (this being a gammal, or IgG1, constant 10 region). The amino acid sequence of RhD2 HC is given as SEQ ID NO: 6. SEQ ID NO: 7 is the nucleotide sequence of the coding region of RhD2 LC. Nucleotides 1-57 encode the signal peptide. Nucleotides 58-388 encode the variable region, of which nucleotides 133-159 encode CDR1, nucleotides 15 211-219 encode CDR2, and nucleotides 328-357 encode CDR3. Nucleotides 389-705 encode the constant region (this being a lambda constant region). The amino acid sequence of RhD2 LC is given as SEQ ID NO: 8. SEQ ID NO: 9 is the nucleotide sequence of the coding region of RhD3 20 HC. Nucleotides 1-57 encode the signal peptide. Nucleotides 58-448 encode the variable region, of which nucleotides 133-162 encode CDR1, nucleotides 214-234 encode CDR2, and nucleotides 349-414 encode CDR3. Nucleotides 449-1578 encode the constant region (this being a gamma3, or IgG3, constant region). The amino acid sequence of RhD3 HC is given as SEQ ID NO: 10. 25 SEQ ID NO: 11 is the nucleotide sequence of the coding region of RhD3 LC. Nucleotides 1-66 encode the signal peptide. Nucleotides 67-391 encode the variable region, of which nucleotides 145-162 encode CDR1, nucleotides 214-222 encode CDR2, and nucleotides 331-360 encode CDR3. Nucleotides 30 392-711 encode the constant region (this being a kappa constant region). The amino acid sequence of RhD3 LC is given as SEQ ID NO: 12. 35 - 24 - WO 20101079510 PCT/IN2009/000741 Alignments of amino acid sequences of RhD1-RhD3 The amino acid sequences of RhD1-RhD3 were aligned with the ClustalW program (www.ebi.ac.uk/Fools/clustalw), using the default parameters 5 from the website. The resulting alignments of HCs and LCs are depicted in Figures 1 and 2, respectively. The variable region of each sequence has been underlined, and the CDRs highlighted in bold (the first occurring CDR, reading the sequences left to right and top to bottom, being CDR1, the second being CDR2, and the third being CDR3). Where the same amino acid occurs in all 10 three chains as aligned, this is identified by a "*" below the relevant amino acid in the bottom sequence (that of RhD3). Similarly, GAP (http://qenome.cs.mtu.edu/aliqn/aliqn.html) using default parameters (Max Match = 11; Min Mismatch = -4; Gap-Open Penalty = 10; Gap 15 Extension Penalty = 2) can be used to align and determine percentage identity between individual pairs of sequences or sections thereof. When so compared, the RhD1 and RhD2 light chain variable regions are 94% identical (104 matches, 6 mismatches, 0 gaps, similarity score of 540), CDR1 regions are 88% identical (8 matches, 1 mismatch, 0 gaps, similarity score of 43), CDR2 regions are 100% 20 identical (3 matches, 0 mismatches, 0 gaps, similarity score of 16), and CDR3 regions are 90% identical (9 matches, 1 mismatch, 0 gaps, similarity score of 43). The RhD1 and RhD2 heavy chain variable regions are 94% identical (123 matches, 7 mismatches, 0 gaps, similarity score of 650), CDR1 regions are 87% identical (7 matches, 1 mismatch, 0 gaps, similarity score of 37), CDR2 regions 25 are 100% identical (8 matches, 0 mismatches, 0 gaps, similarity score of 41), and CDR3 regions are 95% identical (22 matches, 1 mismatch, 0 gaps, similarity score of 131). Expression vectors 30 Two plasmid expression vectors, designated pCB3 and pCB11, were constructed for expressing the antibody heavy and light chains in CHO dhfr cells. - 25 - WO 20101079510 PCT/IN2009/000741 pCB3 This plasmid is illustrated in Figure 3. The components of this plasmid are as listed in Table 2. 5 Table 2 - Components of expression vector pCB3 Vector component Short form Function Source Human Elongation Hua EogainPromoter of Human genomic DNA Factor 1 a with first EFla Promoter intron expression (Clontech) Ampicillin resistance Plasmid propagation Commercial vector AMPr gene (plactamase) in bacteria (pBluescript; Stratagene) Plasmid replication in Commercial vector Origin of replication pUCori bacteria (pBluescript; Stratagene) Simian virus Transcription Commercial vector polyadenylate signal termination (pSV40; BRL/Invitrogen) Simian virus 40E Promoter of Commercial vector SV40E Promoter promoter sequence expression (pSV40; BRL/invitrogen) Bovine growth hormone Transcription Commercial vector BGH Poly(A) triain(Rlnirgn polyadenylate signal termination (BRLInvitrogen) Dihydrofolate reductase DHFR DHFR selection Murine cDNA (Sierra gene marker Biosource, Inc.) 10 pCB1I This plasmid is illustrated in Figure 4. The components of this plasmid are as listed in Table 3. -26- WO 20101079510 PCT/IN2009/000741 Table 3 - Components of expression vector pCB 11 Vector component Short form Function Source Human Elongation EF1a Human genomic DNA Factor 1 a with first Promoter of expression Promoter (Clontech) intron Commercial vector Ampicillin resistance Plasmid propagation in gene (Plactamase) AMPr bacteria (pBluescript; Stratagene) Commercial vector Plasmid replication inComrilvcr Origin of replication pUCori (pBluescript; bacteria Stratagene) Commercial vector Simian virus Transcription polyadenylate signal SV40E poly(A) termination (PSV4O; BRL/Invitrogen) Commercial vector Neomycin Antibiotic selection (pSV-Neo; phosphotransferase neo Mutant BRL/Invitrogen) markerBRfntren (Mutant) modified by Sierra Biosource, Inc. Simian virus 40E SV40E Commercial vector promoter sequence Promoter Promoter of expression (pSV40; BRL/Invitrogen) Bovine growth Bovie grwthTranscription Commercial vector hormone polyadenylate BGH Poly(A) ration Commerleco termination (BRL/lnvitrogen) signal 5 Insertion of recombinant immunoglobulin genes into expression vectors A second PCR was used to amplify the HCs and LCs with appropriate restriction sites added so that the fragments could be inserted into expression vectors. The design of the gene-specific forward primers was based on obtained 10 sequences. The consensus Kozak motif (GCCACC), known to increase the efficiency of eukaryotic translation, was included in each forward primer (Table 5). - 27 - WO 20101079510 PCT/IN2009/000741 The primers (SEQ ID NOs: 20 to 27) for Insertion of RhD1-RhD3 HCs and LCs into expression vectors were as follows. RhD1 HC: 5 Forward gene-specific primer (GSP): 5'-ATCGTCTAGAGCCACCATGGACTGGACCTGGAGGTTCC-3' RhD2 HC: Forward GSP: 10 5'-ATCGTCTAGAGCCACCATGGACTGGACCTGGAGGTTCC-3' RhD3 HC: Forward GSP: 5'-ATCGTCTAGAGCCACCATGGACACACTTTGCTACACACTCC-3' 15 The reverse primer used for all heavy chains: 5'-TGACGAATTCCACTCATTTACCCGGAGACAGG-3' RhD1-RhD2 LCs: 20 Forward GSP: 5'-ATCGTCTAGAGCCACCATGGCCTGGGCTCTGCTATTC-3' Reverse primer: 5'-ACTGGAATTCGAACCTATGAACATTCTGTAGGGG-3' 25 RhD3 LC: Forward GSP: 5'-ATCGTCTAGAGCCACCATGGACATGAGGGTCCCCG-3' 30 Reverse primer: 5'-GACTGAATTCCTAACACTCTCCCCTGTTGAAGC-3' The PCR cycle for insertion of RhD1-RhD3 HCs and LCs into expression vectors comprised the following steps: - 28- WO 20101079510 PCT/IN2009/000741 940C 2min 940C 20s 5 550C 20s 35x 720C 2min (1min for RhD1, RhD2 LC) / 720C 10min 4 0 C hold 10 Construction of IgG3 variant of RhD1 antibody An IgG3 variant of RhD1 was designed as a chimera between the 15 variable region of RhD1 and the constant region of RhD3. The chimerization took advantage of the identical 5' ends of the RhD1 (IgG1) and RhD3 (IgG3) constant regions. The reverse primer specific for variable domain of RhD1 was designed to overlap three codons of the constant region and to introduce silent mutations that created an Nhel restriction site. Identical modification was 20 introduced into the RhD3 constant region 5' end by the forward primer. The Nhel restriction site allowed for convenient in-frame cloning of amplified RhD1 variable domain in front of the RhD3 constant region. This was performed in two steps. First, the constant region of IgG3 HC from RhD3 antibody was amplified, 25 cut with Xbal and EcoRI enzymes, and ligated into Xbal/EcoRI-digested pCB3 vector. In the second step, this intermediary plasmid was re-cut with Xbal and Nhel endonucleases, and the amplified variable region of RhD1, digested with the same enzymes, was inserted. 30 The primers (SEQ ID NO: 28-31) used for Construction of lgG3 variant of RhD were as follows. - 29 - WO 20101079510 PCT/IN2009/000741 Primers used for amplification of RhD3 constant region: Forward: 5'-ATCGTCTAGAGTCAGCTAGCACCAAGGGCCCATCGGTCTTCC-3' 5 Reverse: 5'-TGACGAATTCCACTCATTTACCCGGAGACAGG-3' Primers used for amplification of RhD1 variable domain: 10 Forward: 5'-ATCGTCTAGAGCCACCATGGACTGGACCTGGAGGTTCC-3' Reverse: 5'-GATGCTAGCTGAGGAGACGGTGATCGTGG-3' 15 The PCR cycle for constructing the IgG3 variant of RhD1 comprised the following steps: 940C 2min 20 94 0 C 20s 550C 20s 35x 720C 2min / 720C 10min 25 4 0 C hold PCR enzyme: PfuUltra High-Fidelity thermostable DNA-polymerase (Stratagene). 30 Expression vectors containing cloned antibody genes The RhDI HG, RhD1 LC, RhD2 HC, RhD2 LC, RhD3 HC, RhD3 LC, RhD1V3C HC (chimera composed of the RhD1 heavy chain variable domain and - 30 - WO 20101079510 PCT/IN2009/000741 RhD3 heavy chain constant region) coding sequences as inserted into the expression vectors, including also the added Kozak motifs and restriction sites, are given as SEQ ID NOs: 32, 33, 34, 35, 36, 37, and 38, respectively. Figure 5 is a map of pCB3 illustrating the location of the inserted anti-RhD antibody heavy 5 chain, and Figure 6 is a map of pCB11 illustrating the location of the inserted anti-RhD antibody light chain (the location of insertion being the same, regardless of the specific RhD1, RhD, RhD3 or RhD1V3C heavy or light chain being expressed). 10 Gene optimization Coding sequences of RhD1 and RhD3 antibodies were optimized by GENEART AG using proprietary algorithms. The optimized coding sequences for RhD1 HC, RhD1 LC, RhD3 HC, and RhD3 LC are given as SEQ ID NOs: 39, 15 40, 41 and 42, respectively. Cloning of optimized RhD1 genes into expression vectors The optimized genes for RhD1 were subcloned into pCB expression 20 vectors. To add the restriction sites necessary for cloning, the coding regions were amplified by PCR using the primers listed below. Each amplified fragment was inserted in the respective vector and verified by sequencing. The primers (SEQ ID NOs: 43 to 46) that were used for appending the 25 restriction sites compatible with the pCB expression vectors to the optimized RhD1 genes are as follows. Optimsed RhD1 HC: Forward: 30 5'-ATCGTCTAGAGCCACCATGGACTGGACCTG-3' Reverse: 5'-ATCGGGATCCTCATCACTTGCCGGGGGAC-3' - 31 - WO 20101079510 PCT/IN2009/000741 Optimised RhD1 LC: Forward: 5'-ATCGTCTAGAGCCACCATGGCCTGGGCCC-3' 5 Reverse: 5'-ATCGGGATCCTCATCAGCTGCACTCGGTGGGG-3' The Xbal and BamHl sites in the primers are underlined. 10 The optimized RhD1 HC and RhD1 LC coding sequences as inserted into the expression vectors, including added Kozac motifs and restriction sites, are given as SEQ ID NOs: 47 and 48. Cell culture 15 Growth media MEM. growth medium was used at all stages of recombinant CHO cell line development work. The components, formulation, and material sources are shown in Table 4. After the addition of all components, the complete medium 20 was filtered through a 0.22 lam filter (Stericup-GP 0.22 lam filter unit, Millipore or equivalent). - 32 - WO 20101079510 PCT/IN2009/000741 Table 4 - Culture media Medium Components Vendors Catalog # Final concentration CHO DXB11 MEMa Gibco or 32561-037 or 1x Host Cell without Cellgro CV2561-049 1x Growth ribonucleosides and Medium 1 deoxyribonucleosides HT, 250x Gibco 31985-070 1x Gamma-irradiated Hyclone SH30079.33 7.5% dialyzed fetal bovine serum (dFBS) GlutaMax, 100x Gibco 35050-061 1x CHO DXB11 MEMa Gibco 32571-036 1x Host Cell with ribonucleosides Growth and Medium 2 deoxyribonucleosides Gamma-irradiated Hyclone SH30070.03 7.5% fetal bovine serum (FBS) GlutaMax, 100x Gibco 35050-061 1x Transfectant MEMa Gibco or 32561-037 or 1x Selection without Cellgro CV2561-049 1x Medium ribonucleosides and deoxyribonucleosides. Gamma-irradiated Hyclone SH30079.33 7.5% dFBS GlutaMax, 100x Gibco 35050-061 1x Geneticin (a G-418 Gibco 10131-027 0.5 mg/ml formulation) 5 Freezing media The composition of the freezing media used for cryopreservation of cells is given in Table 5. - 33 - WO 20101079510 PCT/IN2009/000741 Table 5 - Components of freezing media Freezing medium 1: Components Vendors Catalog # Volume per 100 ml Gamma-irradiated HyClone SH30079.33 95 mL dFBS dimethyl sulfoxide Sigma D2438 5 mL (DMSO) 5 Freezing medium 2: Components Vendors Catalog # Volume per 100 ml Gamma-irradiated HyClone SH30070.03 90 mL FBS DMSO Sigma D2438 10 mL Maintenance of cells 10 Dihydrofolate reductase (DHFR)-deficient CHO DXB1 1 cells were grown in Host Cell Growth Medium 1 or 2 (Table 4) and were split every 3-4 days. Cell density and viability measurements 15 Viable cell density and cell viability was determined using the Trypan Blue exclusion method and a hemocytometer (Hausser Scientific). Stable transfection and amplification in methotrexate (MTX) 20 CHO DXB1 1 cells were co-transfected with equal amounts of plasmid DNA coding for the light and heavy chains of the human IgG (Table 6). Transfections were performed using Lipofectamine 2000 reagent (Invitrogen) -34- WO 20101079510 PCT/IN2009/000741 following the manufacturer's recommendation. Stable transfectants were selected using Transfectant Selection Medium (Table 4). Table 6 - Conditions for a typical transfection of CHO DXB 11 cells 5 Vessel Amount of HC Amount of LC Amount of DNA DNA Lipofectamine 2000 T75 flask or 15pg 15pg 30-75l 10cm dish Transfected cells were cultured for 2 days at 370C and 5% C02 in Host Cell Growth Medium 1 or 2 prior to initiation of the selection process by replacing 10 the Growth Medium with Transfectant Selection Medium (Table 4). During the selection process, the spent medium was removed and replaced with fresh medium whenever necessary. After the selection process was completed and the transfected cells resumed growing, the cells were either 15 -- transferred into the Transfectant Selection Medium (Table 4) containing various levels of MTX (Calbiochem) for amplification of antibody genes, or -- subcloned (see below). In this case, 12 best-producing clones were selected and pooled for further amplification in MTX. 20 Single cell cloning In order to select single-cell clones, stably transfected cells were plated in an appropriate number of flat-bottom 96-well plates at 0.5-1 cell per well. During the process, the cell growth and health was monitored under the microscope. 25 Cells were cultured for approximately two weeks prior to selection of the best producing clones by screening with ELISA. - 35 - WO 20101079510 PCT/IN2009/000741 Enzyme-linked Immunosorbent Assay (ELISA) The antibody titers during all stages of cell line development were evaluated with the Human IgG ELISA Quantitation Kit (Bethyl Laboratories) 5 according to manufacturer's instructions. Shortly, the Nunc Maxisorp ELISA plates were coated with Fc-specific goat anti-human IgG polyclonal antibody in phosphate-buffered saline (PBS). Plates were incubated overnight at 4'C. Next day, the plates were washed three times and blocked for 1 hour with powdered non-fat milk dissolved in the wash buffer. After a washing step, samples and 10 standards were pipetted onto the plates and incubated at room temperature for 1 hour, followed by three washes. Secondary antibody conjugated to horseradish peroxidase (HRP) was then added to each well and the plates were incubated again at room temperature for 1 hour. Plates were washed three times with wash buffer, rinsed once with distilled water, and tapped dry. 15 Tetramethylbenzidine (TMB)-containing substrate was added to each well and color was allowed to develop for 15 minutes at room temperature. The reaction was stopped by sulfuric acid and the plates were read on a plate reader (Bio Rad, Molecular Dynamics, or Dynex Technologies) at 450nm. The data was analyzed with a software package supplied with the plate reader. 20 Expression of recombinant antibodies from cell pools stably transfected with non-optimized cDNAs The scheme of transfections (performed according to Table 6) and 25 designations of the transfected cells are provided in Table 7. Table 7 - Designated name for transfected pools. HC DNA in pCB3 LC DNA in pCB1 1 Name of Stable Recombinant IgG Pool Isotype RhD1 gamma RhD1 lambda RhD1 IgGI RhD1V3C gamma RhD1 lambda RhD4 IgG3 RhD2 gamma RhD2 lambda RhD2 IgG1 RhD3 gamma RhD3 kappa RhD3 IgG3 - 36 - WO 2010/079510 PCT/IN2009/000741 Generally, a better expression was reached when the transfected cells were subcloned after the selection process, the clones were ranked for antibody production by ELISA, and only the pools of 12 best-producing clones were amplified in MTX. Amplification of selected but non-subcloned transfectants 5 yielded pools exhibiting lower productivity, albeit in shorter time. One typical scheme of MTX amplification is shown below: -- Selected cells (OnM MTX) were transferred in parallel to Transfectant Selection Medium containing 50nM or 100nM MTX (Step 1) -- Cells recovered from Step 1 were expanded and split into 200nM and 10 500nM MTX (Step 2) -- Cells that have survived Step 2 were expanded and subjected to amplification in 1000nM MTX (Step 3) At each step, the antibody productivity was assessed by ELISA (Table 8). 15 Table 8 - Examples of productivity of unamplified and amplified pools of 12 best clones Pools of 12 best Antibody expression levels clones MTX level (nM) after 7 day culture (Vig/ml) RhD1 0 10.8 RhD1 50 5.66 RhD1 200 6.44 RhD1 500 9.12 RhD1 1000 27.8 RhD2 0 9.25 RhD2 50 12.25 RhD2 100 12.75 RhD2 200 18.4 RhD3 0 4.08 -37- WO 2010/079510 PCT/IN2009/000741 RhD3 200 3.14 RhD3 500 6.85 RhD4 0 1.2 RhD4 0 2 The pools yielding the best antibody titers were expanded in tissue culture flasks in Transfectant Selection Medium (without MTX and Geneticin and 5 containing low bovine igG FBS instead of regular FBS). The supernatants from these cultures were collected and used for purification of the antibodies. Expression of RhD1 and RhD3 antibodies by transfected and amplified 10 clonal cell populations adapted to serum-free media As the levels of antibody expression obtained from the cell pools (Table 8) were still not as high as desired, the transfection, selection and amplification process was carried out anew, this time employing a subcloning step (as 15 described above) after each amplification step, in addition to after the initial selection step, so as to obtain clonal cell lines (single cell clones) expressing amplified levels of anti-RhD antibody. More specifically, CHO DXB1 1 cells were transfected with plasmids 20 encoding the heavy and light chains of either RhD1 or RhD3. Transfection and selection of stably transfected cells was carried out in essentially the same manner as described above. Transfected cells were then subcloned, and the resulting clones screened for antibody production. The most productive clonal cell lines were amplified. After amplification, the cells were again subcloned, and 25 the most productive clones subjected to a further round of amplification and subcloning. The selection media, and the amplification media used for the first and second amplification steps, are listed in Table 9. - 38 - WO 20101079510 PCT/IN2009/000741 The final best producing clonal cell lines (obtained after both rounds of amplification) were adapted to suspension growth in commercial serum-free media (IS CHO_CD4TM, Irvine Scientific). This task was performed either in the shake flasks or in spinner bottles by seeding the cells in a 1:1 mixture of the final 5 amplification media (Table 9) and a serum-free media containing the same level of MTX, and then gradually increasing the proportion of the serum-free media over a period of four to six weeks until the cells were fully capable of growing in 100% serum-free medium. 10 The maximum productivities of the best producing RhD1 and RhD3 clonal cells lines, before and after the adaptation to serum-free media, are listed in Table 9. The supernatants from these cultures were again collected and used for purification of the antibodies. - 39 - WO 20101079510 PCT/IN2009/000741 Table 9 - Selection and amplification media for five selected RhD clones. Included are productivity data before and after the adaptation to serum-free media. RhD1 RhD1 RhD3 RhD3 Recombinant Clone: Clone 1 Clone 6 Clonel Clone 4 Gene Optimization: Yes Yes No No Transfectant Transfectant Transfectant Transfectant Selection Selection Selection Selection: Selection Selection Medium Medium and Medium Medium 2AnM MTX 2nM MTX Amplification Transfectant Transfectant Transfectant Transfectant Media. Selection Selection Selection Selection The Amplification Medium Medium Medium Medium composition Step 1: No G418 No G418 No G418 No G418 of the 300nM MTX 300nM MTX 200nM MTX 200nM MTX Transfectant Transfectant Transfectant Selection Transfectant Transfectant Selection Selection Medium is Selection Selection Amplification Medium Medium listed in Table Medium Medium Step 2: No G418 No G418 4. No G418 No G418 2,400nM 1 ,200nM 800nM MTX 800nM MTX MTX MTX Before adaptation to serum-fre t87 g/ml 100 g/ml 128 g/ml 87 pg/mi serum-free Antibody media Productivity After adaptation to serum-fre t419 pg/ml 431 pg/ml 320 ig/ml 326 ig/ml serum-free media 5 -40- WO 2010/079510 PCT/IN2009/000741 Antibody purification The pH of the culture supernatants was adjusted to pH 7.2 with 1N NaOH. Each supernatant was filtered through a 0.
2 p filter and loaded on a 5 protein A column pre-equilibrated in phosphate-buffered saline (PBS). The column was washed with PBS to remove all the unbound material from the culture supernatant. The antibody bound to the protein A column was eluted with 0.1 M Glycine (pH 2.5). The eluate was neutralized with 2M Tris buffer adjusted to pH 8.0. The eluate containing monoclonal antibody was dialyzed against 10 PBS. The anti-RhD antibody concentration was determined by agglutination assay using D positive erythrocytes. The antibody concentration was determined spectrophotometrically at 280 nm using an optical density value of 1.4 OD for a 1 mg/ml solution based on the molar extinction coefficient for human monoclonal antibody. 15 Anti-D quantitation by hemagglutination assay The anti-RhD antibody levels in. the supernatants and purified antibody 20 were quantified by measuring the agglutination of bromelain-treated RhD positive erythrocytes using the Technicon Autoanalyzer system as previously described by Gunson et. al (H. H. Gunson, P. K. Phillips, and F. Stratton J. clin. Path., 1972, 25, 198-205. Polyclonal Anti-RhD antibodies from NIBSC ( 2 nd International standard 01/572) were used as a standard. 25 Briefly, bromelain-treated RhD positive red cells are incubated with various concentrations of anti-RhD antibodies. The cells are allowed to agglutinate over a period of time. The agglutinated cells are removed in the autoanalyzer and the rest of the erythrocytes are lysed using detergent. The 30 optical density of the released hemoglobin is measured spectrophotometrically. The anti-D concentrations of the samples are calculated using a standard graph obtained from various concentrations of the Anti-D standard. -41- WO 2010/079510 PCT/IN2009/000741 Flow cytometry assay Each human anti-RhD monoclonal antibody was serially diluted 1 in 3 down from 0.5 mg/mI to prepare the total of 15 dilutions. Each dilution was 5 added to 1-5x10 5 RhD positive or RhD negative human red blood cells (RBCs), with otherwise matching genotypes, pretreated with papain to make the antigenic components of RhD more accessible to the antibodies. An anti-human IgG antibody labelled with Fluorescein Isothiocyanate (FITC) was used as a secondary antibody to stain antibodies bound to the RBCs. 10 The samples were analyzed on the FACSort instrument (Becton Dickinson). The RBC population was gated for based on the forward- and side scatter parameters. Fluorescence of RhD negative samples was considered a background, since these cells lack the RhD antigen that is targeted by anti-RhD 15 antibodies. RhD negative cells incubated with a particular concentration of antibody therefore served as a negative control for RhD positive cells incubated with the same antibody dilution. The specific fluorescence and the percentage of RhD positive cells bound by anti-RhD antibody (and stained with FITC labelled anti-human IgG) was then determined, for each dilution of anti-RhD antibody, 20 based on the difference between the level of fluorescence in the RhD positive and RhD negative samples. For each anti-RhD antibody, the percentage of positive cells bound by the antibody was plotted against the logarithm of the antibody concentration, and EC50 was estimated from this chart. This provided basic information about the binding affinity and specificity of the antibodies for 25 the RhD antigen. ADCC assay The effectiveness of the anti-RhD antibodies in eliminating RhD-positive 30 red blood cells in vivo, and thus utility of the antibodies in preventing immunization of an RhD-negative individual exposed to RhD-positive blood, was gauged via an antibody-dependant cellular toxicity (ADCC) assay. The ADCC assay was based on the method described by Miescher et. al. 35 (British Journal of Haematology 2000 111:157-166). RhD positive erythrocytes - 42 - WO 2010/079510 PCT/IN2009/000741 were treated with papain and subsequently labeled with the fluorescent dye 5 (and 6) carboxyfluorescein diacetate succinimidyl ester. The labeled erythrocytes were preincubated with varying concentrations (0.1-50 ng/ml) of anti-RhD antibodies for 1 hr. Peripheral blood mononuclear cells (PBMCs) were 5 added to the erythrocyte suspension and incubated for 18 hrs in a C02 incubator at 37 0 C. The extent of the target cell lysis at the end of incubation was determined by measuring the release of the dye from lysed RBCs into supernatant with a fluorometer. The percentage of cytotoxicity was calculated according to the following formula: 10 Fcexp - Fcmed % specific lysis = x 100 Fcde, - Fcmed 15 where Fcexp = fluorescence of samples Fcdet = maximum fluorescence control (obtained by lysing the RBCs with a detergent (1% Triton-X1 00)) Fcmed = background fluorescence control (spontaneous release of the dye from 20 RBCs in the absence of PBMCs and antibody) The percentage of cytotoxicity was then plotted against the logarithm of antibody concentration at which the erythrocytes were preincubated, and this data used to calculating the EC50, i.e. the effective concentration of antibody 25 causing 50% of the maximum specific lysis achievable by that antibody. By way of example, Fig. 7 is a plot of percentage cytotoxicity again antibody concentration generated from the results of an ADCC assay using an NIBSC standard (anti-RhD polyclonal antibodies). This dose-response dependence theoretically yields a sigmoid curve with a near-linear middle region. To perform 30 a linear approximation in this region, a straight line can be fitted to the pertinent data points by linear regression using a suitable software package (such as, for example, Microsoft Excel
TM
). Fig. 8, for example, is a linear regression performed on the relevant data points from Fig. 7. An equation representing this straight line can then be used to calculate the EC50. For example for the data in -43- WO 2010/079510 PCT/IN20091000741 Fig. 7, where the maximum specific lysis caused by the NIBSC standard polyclonal antibody was approximately 88% compared to the detergent-induced lysis (100%), the EC50 was calculated for the value of specific lysis equalling 44%. 5 Hemagglutination and ADCC assay results Results of hemagglutination and ADCC assays, carried out in accordance with the procedures described above, are shown below in Table 10. 10 Agglutination titers are expressed as micrograms of active (RhD antigen binding) antibody per mg of protein. The EC50 values were determined from two independent experiments. Table 9 - Agqlutination titers and EC50 values for RhD1, RhD3, and RhD4 15 antibodies. A control polyclonal antibody (NIBSC Standard) and two batches of a control monoclonal antibody are included for comparison. Agglutination ADCC EC50 (ng of Active Ab/mi) Antibody Titer (tg of Active Ab per mg of Protein) Experiment I Experiment 2 Average Control anti-RhD mAb Batch No. 1 100.0 1.2 2.1 1.7 Control anti-RhD mAb Batch No. 2 100.0 0.9 1.9 1.4 NIBSC Standard (anti-RhD Polyclonal Ab) 7.1 0.5 1.3 0.9 RhD1 Clone 1 716.2 0.7 1.5 1.1 RhD1 Clone 6 378.1 0.4 0.9 0.7 RhD3 Clone 1 324.3 0.2 0.3 0.3 RhD3 Clone 4 275.3 0.1 0.2 0.2 RhD4 303.3 0.1 0.5 0.3 -44- WO 20101079510 PCT/IN2009/000741 Formulations The purified monoclonal anti-RhD antibodies can be formulated for administration via any suitable route. Typically, the antibodies are administered 5 via injection. In such circumstances, the antibody is typically formulated as a liquid suspension of the antibodies in a suitable buffer solution. Exemplary buffers include: phosphate-buffered saline (20 mM phosphate buffer (pH 6.8) containing 150 mM NaCI); and 10 glycine saline buffer (0.3 M glycine containing 0.15 M NaCl adjusted to pH 6.5). Preferred formulations comprise both monoclonal antibodies having an IgG 1 constant region and monoclonal antibodies having an IgG 3 constant 15 region. Thus, formulations comprising RhD1 antibodies (which are of the IgG 1 isotype) in combination with RhD3 antibodies (which are of the IgG 3 isotype) and/or RhD4 antibodies (which consist of the RhD1V3C heavy chain and RhD1 light chain) are preferred. -45-

Claims (20)

1. An isolated anti-RhD monoclonal antibody comprising: a) a heavy chain variable region having first, second and third CDRs 5 which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 2, and a light chain variable region having a first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 4; or 10 b) a heavy chain variable region having first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 6, and a light chain variable region having first, second and third CDRs which are identical or substantially identical to the respective first, 15 second, and third CDRs of SEQ ID NO: 8; wherein two CDRs are substantially identical if they have amino acid sequences that are at least 80% identical and/or differ in no more than one amino acid. 20
2. The antibody of claim 1, comprising: a) a heavy chain variable region which is at least 80% identical to the variable region of SEQ ID NO: 2 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 2, and 25 a light chain variable region which is at least 80% identical to the variable region of SEQ ID NO: 4 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 4; or b) a heavy chain variable region which is at least 80% identical to the 30 variable region of SEQ ID NO: 6 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 6, and a light chain variable region which is at least 80% identical to the variable region of SEQ ID NO: 8 and has first, second and third - 46 - CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 8.
3. The antibody of claim 2, wherein the antibody comprises: a heavy chain 5 variable region which is at least 80% identical to the variable region of SEQ ID NO: 2, and has first, second and third CDRs which are identical to the respective first, second and third CDRs of SEQ ID NO: 2; and a light chain variable region which is at least 80% identical to the variable region of SEQ ID NO: 4, and has first, second and third CDRs which are 10 identical to the respective first, second and third CDRs of SEQ ID NO: 4.
4. The antibody of claim 2, wherein the antibody comprises: a heavy chain variable region which is at least 80% identical to the variable region of SEQ ID NO: 6, and has first, second and third CDRs which are identical 15 to the respective first, second and third CDRs of SEQ ID NO: 6; and a light chain variable region which is at least 80% identical to the variable region of SEQ ID NO: 8, and has first, second and third CDRs which are identical to the respective first, second and third CDRs of SEQ ID NO: 8. 20
5. The antibody of any one of claims 2 to 4, wherein the respective variable regions are at least 90% identical.
6. The antibody of claim 5, wherein the respective variable regions are at least 95% identical. 25
7. The antibody of claim 6, wherein the respective variable regions are identical.
8. The antibody of any preceding claim, wherein the antibody comprises a 30 light chain constant domain and a heavy chain constant domain.
9. The antibody of claim 8, wherein the antibody comprises a heavy chain constant region. - 47 -
10. The antibody of claim 8 or 9, wherein said heavy chain constant domain or region is an IgG 1 or IgG 3 constant domain or region.
11. An isolated polynucleotide encoding the light and/or heavy chain of an 5 antibody according to any one of the preceding claims.
12. An expression vector including coding sequences encoding the light and heavy chains of an antibody according to any one of claims 1 to 10. 10
13. An expression system including coding sequences encoding the light and heavy chains of an antibody according to any one of claims 1 to 10, the expression system comprising: a first expression vector including the coding sequence encoding the light chain; and 15 a second expression vector including the coding sequence encoding the heavy chain.
14. A cell transformed with an expression vector of claim 12 or the expression system according to claim 13. 20
15. A method of manufacturing monoclonal antibodies, comprising cultivating recombinant cells according to claim 14, and recovering the monoclonal antibody from the culture medium. 25
16. A pharmaceutical composition comprising a monoclonal antibody according to any one of claims 1 to 10, the isolated polynucleotide according to claim 11, the expression vector according to claim 12, the expression system according to claim 13 or the cell according to claim 14. 30
17. The pharmaceutical composition of claim 16, wherein the pharmaceutical composition comprises a first monoclonal antibody according to any one of claims 1 to 10, and a second monoclonal antibody any one of claims 1 to 10, wherein said first and second monoclonal antibodies are distinct from one another. - 48 -
18. The pharmaceutical composition of claim 17, wherein the first monoclonal antibody has a heavy chain comprising an IgG 1 constant domain or region, and the second monoclonal antibody has a heavy chain 5 comprising an IgG 3 constant domain or region.
19. A method of inhibiting or preventing immunization of a RhD-negative human patient against RhD-positive blood, comprising administering a prophylaxis effective amount of a monoclonal antibody according to any 10 one of claims 1 to 10, or a pharmaceutical composition according to any one of claims 16 to 18.
20. Use of a monoclonal antibody according to any one of claims 1 to 10, the isolated polynucleotide according to claim 11, the expression vector 15 according to claim 12, the expression system according to claim 13 or the cell according to claim 14 in the manufacture of a medicament for inhibiting or preventing immunization of a RhD-negative human patient against RhD-positive blood. - 49 -
AU2009336567A 2008-12-31 2009-12-24 Anti-RhD monoclonal antibodies Active AU2009336567B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2014200308A AU2014200308B2 (en) 2008-12-31 2014-01-17 Anti-RhD monoclonal antibodies

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
IN2730/MUM/2008 2008-12-31
IN2730MU2008 2008-12-31
PCT/IN2009/000741 WO2010079510A2 (en) 2008-12-31 2009-12-24 Anti-rhd monoclonal antibodies

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2014200308A Division AU2014200308B2 (en) 2008-12-31 2014-01-17 Anti-RhD monoclonal antibodies

Publications (2)

Publication Number Publication Date
AU2009336567A1 AU2009336567A1 (en) 2011-06-30
AU2009336567B2 true AU2009336567B2 (en) 2014-02-20

Family

ID=42316926

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2009336567A Active AU2009336567B2 (en) 2008-12-31 2009-12-24 Anti-RhD monoclonal antibodies

Country Status (10)

Country Link
US (1) US8529903B2 (en)
EP (2) EP2450381B1 (en)
CN (1) CN102272160A (en)
AU (1) AU2009336567B2 (en)
CA (1) CA2746350C (en)
EA (1) EA024722B1 (en)
EC (1) ECSP11011190A (en)
ES (2) ES2740051T3 (en)
MX (1) MX2011006869A (en)
WO (1) WO2010079510A2 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102180660B1 (en) * 2013-05-03 2020-11-19 후지필름 다이오신쓰 바이오테크놀로지스 유케 리미티드 Expression process
WO2015095002A1 (en) * 2013-12-16 2015-06-25 Texas Tech University System Anti-ron monoclonal antibodies as a cytotoxic drug delivery system for targeted cancer therapy
SI3126391T1 (en) 2014-03-31 2020-04-30 Universitetet I Tromso - Norges Arktiske Universitet Antibodies against hpa-1a
CA2956072A1 (en) 2014-07-21 2016-01-28 Bloodworks Antibodies that recognize red blood cell antigens
WO2017005923A1 (en) * 2015-07-09 2017-01-12 Institut National De La Sante Et De La Recherche Medicale (Inserm) Lentiviral vector expressing membrane-anchored or secreted antibody
CN114409791B (en) * 2022-01-26 2023-01-24 南京医科大学 Fully human anti-human erythrocyte RhD full molecular IgG and preparation method and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996007740A1 (en) * 1994-09-02 1996-03-14 Institut Pasteur Monoclonal recombinant anti-rhesus d (d7c2) antibody

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8610106D0 (en) 1986-04-25 1986-05-29 Central Blood Lab Authority Human igm-producing heterohybridoma
GB8722018D0 (en) 1987-09-18 1987-10-28 Central Blood Lab Authority Human anti-rh(d)monoclonal antibodies
US6475787B1 (en) 1989-07-28 2002-11-05 Wyeth Method for producing monoclonal antibodies
FR2807767B1 (en) 2000-04-12 2005-01-14 Lab Francais Du Fractionnement MONOCLONAL ANTIBODIES ANTI-D

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996007740A1 (en) * 1994-09-02 1996-03-14 Institut Pasteur Monoclonal recombinant anti-rhesus d (d7c2) antibody

Also Published As

Publication number Publication date
AU2009336567A1 (en) 2011-06-30
US8529903B2 (en) 2013-09-10
MX2011006869A (en) 2011-07-19
ECSP11011190A (en) 2011-09-30
CA2746350A1 (en) 2010-07-15
CN102272160A (en) 2011-12-07
CA2746350C (en) 2018-06-12
WO2010079510A3 (en) 2010-10-07
EP2370469A2 (en) 2011-10-05
EA024722B1 (en) 2016-10-31
EP2450381A1 (en) 2012-05-09
ES2740051T3 (en) 2020-02-05
EP2450381B1 (en) 2019-07-03
ES2692173T3 (en) 2018-11-30
EP2370469B1 (en) 2018-09-05
WO2010079510A2 (en) 2010-07-15
EA201100884A1 (en) 2012-02-28
US20120027769A1 (en) 2012-02-02

Similar Documents

Publication Publication Date Title
JP4926054B2 (en) Anti-RhD recombinant polyclonal antibody and production method
JP6124958B2 (en) Anti-D monoclonal antibody
AU2009336567B2 (en) Anti-RhD monoclonal antibodies
CA2722348A1 (en) Methods for manufacturing a polyclonal protein
Miescher et al. CHO expression of a novel human recombinant IgG1 anti‐RhD antibody isolated by phage display
AU2014200308B2 (en) Anti-RhD monoclonal antibodies
WO2018078643A1 (en) Monoclonal antibodies neutralizing the tetanus toxin
CN116284358A (en) Neutralizing antibody specifically binding to novel coronavirus or mutant spike protein NTD epitope thereof, and preparation method and application thereof

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)